Hepatic Mechanisms of Action in the Triglyceride-Lowering Effect of Omega-3 Fatty Acid-Based Therapies

Last Updated: August 05, 2022


Disclosure: No relevant disclosures.
Pub Date: Monday, Aug 19, 2019
Author: Joanne Hsieh, PhD
Affiliation: Columbia University Irving Medical Center

The atherosclerotic cardiovascular disease (ASCVD) risk posed by hypertriglyceridemia has become clear with Mendelian randomization studies, which use genetic variants in the human population to naturally assign individuals to a “control” or “treatment” group. Single nucleotide polymorphisms in loci such as APOC3 and ANGPTL3 that lower plasma triglycerides (TG) are significantly associated with CVD risk.1 Moreover, exome sequencing studies that examine protein coding variants in these genes have also indicated a protective effect of lowering triglycerides.2 However, the promising therapies targeting apolipoprotein C-III and angiopoietin-like 3 are currently monoclonal antibodies or antisense oligonucleotides that are not only expensive, but need to be administered by injection.3-5 Pharmacotherapies based on n-3 fatty acids (FA) present orally-available options that are relatively affordable. As detailed in the AHA Science Advisory by Skulas-Ray et al., n-3 FA-based therapies are also efficacious in lowering triglycerides in hypertriglyceridemic individuals.

The main mechanism of action for n-3 FA-based therapies is likely the inhibition of sterol regulatory element binding protein-1 (SREBP-1), the main transcriptional activator of de novo lipogenesis (DNL) (synthesis of new fatty acids). In humans with non-alcoholic fatty liver disease (NAFLD), which is considered the hepatic component of the metabolic syndrome, DNL is increased more than 3-fold,6 indicating excessive SREBP-1 activity. SREBPs are synthesized as a precursor form that is a polytopic membrane protein that interacts with SREBP cleavage-activating protein (SCAP) and is retained in the endoplasmic reticulum (ER) by another transmembrane protein, insulin-induced gene (INSIG). Upon insulin stimulation, SCAP escorts SREBP to the Golgi apparatus where it undergoes proteolytic cleavages to yield a soluble domain that translocates to the nucleus to direct lipogenic gene expression.7

Studies in cell culture and rodents have proposed multiple molecular mechanisms for how polyunsaturated fatty acids (PUFAs) of multiple species inactivate SREBP-1.8-10 The ER-localized ubiquitin-like domain-containing protein d8 (Ubxd8) polymerizes in response to unsaturated fatty acids, which protects INSIG from ER-associated degradation.11 The stabilization of INSIG helps to sequester SREBP-1 in the ER, where it cannot be processed to its transcriptionally active form. Such a mechanism is also expected to affect SREBP-2, which directs the synthesis of cholesterol and increases low density lipoprotein receptor (LDLR) expression. However, n-3 FAs have repeatedly been shown to be specific for SREBP-1 activation. One group reported that docosahexaenoic acid (DHA) activates ERK, which leads to the phosphorylation of nuclear SREBP-1. Phosphorylation accelerates the turnover of nuclear SREBP-1 by proteasomal degradation, thus limiting the activation of lipogenic genes. Interestingly, the protein stability of nuclear SREBP-1 is much more sensitive to DHA than eicosapentaenoic acid (EPA).12 Both DHA and EPA have been documented to reduce Srebf1 mRNA,10,12 possibly because SREBP-1 drives the expression of its own transcript.8,13 Figure 1 pictorially depicts the mechanisms by which n-3 FA can inhibit SREBP-1 activation.

Figure 1. Inhibition of SREBP-1 activity by n-3 FA. The effects of n-3 FA are shown in red symbols. The precursor form of SREBP-1 (orange) is retained along with SCAP (green) in the ER by INSIG (purple). Unsaturated FA promotes Ubxd8 (yellow) polymerization in the ER, which prevents it from mediating the proteasomal degradation of INSIG. Additional mechanisms not yet elucidated by inhibit the transport of SREBP-1/SCAP from the ER to the Golgi apparatus. In the Golgi, SREBP-1 is processed by two proteases (blue), which releases the nuclear (N) domain that directs transcription in the nucleus. DHA promotes the proteasomal degradation of the nuclear form of SREBP-1. In addition to lipogenic genes, SREBP-1 promotes the expression of its own transcript. Therefore, by these various mechanisms that act to suppress the amount of nuclear SREBP-1, overall hepatic levels of SREBP-1 levels also decrease.

These findings in rodents appear to be translatable to humans. In subjects taking 4 g/day EPA + DHA, who had >2% DHA enrichment in their erythrocytes (a reliable surrogate of hepatic DHA enrichment), the fraction of DNL-derived TG in the very low density lipoprotein (VLDL) fraction was halved.14 However, despite the elevated DNL rates in patients with insulin resistance, exogenous FA derived from lipolysis of adipose stores or chylomicrons continue to comprise the majority of VLDL-TG.15 An important SREBP-1 transcriptional target is glycerol-3-phosphate acyltransferase 1 (GPAT1), which catalyzes the first committed step in hepatic TG synthesis using exogenous FA. In mice, DHA supplementation to suppress SREBP-1 activity decreased Gpat1 expression 3-fold and normalized hypertriglyceridemia.16

The reason for increased LDL-cholesterol with some n-3 FA formulations in hypertriglyceridemic individuals remains unclear. As alluded to earlier, n-3 fatty acids have never been demonstrated in vivo to affect the activation of SREBP-2, which drives the expression of LDLR and proprotein convertase subtilisin/kexin type 9 (PCSK9). PUFAs have been shown in vitro to bind the oxysterol sensor liver X receptor (LXR),17 which could possibly raise LDL-cholesterol by lowering ATP-binding cassette G5/8 (ABCG5/8) expression and therefore biliary cholesterol excretion, but this has not been shown to occur in vivo.18 DHA has been shown to be a ligand for the bile acid nuclear receptor farsenoid X receptor (FXR).19 The FXR agonist obeticholic acid has been reported to increase LDL-cholesterol in humans,20 which contradicts the findings in rodent models. Mice also have a poor response to statins, which precludes any detailed mechanistic studies on the lipoprotein effects of n-3 FA in the setting of statin treatment, even though it would be more reflective of the patient population.

All of the above-described molecular mechanisms can be ascribed to both DHA and EPA, so it is puzzling that EPA-only formulations such as icosapentyl ethyl esters (IPE) seem to have a different effect on LDL-cholesterol. However, it may be premature to conclude that EPA-only formulations are better for avoiding increases in LDL-cholesterol. The MARINE, ANCHOR, and REDUCE-IT trials on IPE all used light liquid paraffin (mineral) oil as the placebo control. Mineral oil is known to block the absorption of many medications, including statins. Consistent with this effect, increases in the particle concentration of most apolipoprotein B-containing lipoproteins were observed in the placebo group of the MARINE trial.21 Interestingly, when the subjects were split up according to statin treatment, there was a trend for an enhanced LDL-lowering effect of IPE in those taking statins.21 While the statin-treated groups in MARINE were small, this observation is suggestive of an effect of compromised statin absorption in the placebo group. Tellingly, the open-label JELIS trial that did not use a mineral oil control reported no differences in LDL-C.22 Therefore, the results expressed as placebo-adjusted values may not be a true reflection of EPA-only formulations’ effect on LDL-C.

In addition to improving hypertriglyceridemia, treatment with n-3 FA-based therapies may lower ASCVD incidence by addressing another risk factor. A recent large meta-analysis suggests that NAFLD is an independent risk factor for CVD, even after adjusting for traditional risk factors including body mass index, smoking and LDL-cholesterol.23 The WELCOME trial on Lovaza showed demonstrate that 4 g/day EPA + DHA was efficacious in reducing liver fat.24 A sub-study of the WELCOME trial reported a direct relationship between change in liver fat percentage and carotid intima-media thickness (CIMT) progression. There was also a direction relationship between a marker of hepatic necroinflammation and CIMT progression. Both of these correlations were independent predictors of CIMT progression even after adjusting for plasma TG concentrations.25 The use of n-3 FA-based therapies may therefore yield benefits in addition to plasma TG lowering in the patient who is at risk of ASCVD.

Citation


Skulas-Ray AC, Wilson PWF, Harris WS, Brinton EA, Kris-Etherton PM, Richter CK, Jacobson TA, Engler MB, Miller M, Robinson JG, Blum CB, Rodriguez-Leyva D, de Ferranti SD, Welty FK, on behalf of the American Heart Association Council on Arteriosclerosis, Thrombosis and Vascular Biology, Council on Lifestyle and Cardiometabolic Health, Council on Lifelong Congenital Heart Disease and Heart Health in the Young, Council on Cardiovascular and Stroke Nursing, and Council on Clinical Cardiology. Omega-3 fatty acids for the management of hypertriglyceridemia: a science advisory from the American Heart Association [published online ahead of print August 19, 2019]. Circulation. doi: 10.1161/CIR.0000000000000709.

References


  1. Do R, Willer CJ, Schmidt EM, Sengupta S, Gao C, Peloso GM, et al. Common variants associated with plasma triglycerides and risk for coronary artery disease. Nat Genet. 2013,45:1345-52.
  2. Liu DJ, Peloso GM, Yu H, Butterworth AS, Wang X, Mahajan A, et al. Exome-wide association study of plasma lipids in >300,000 individuals. Nat Genet. 2017,49:1758-1766.
  3. Dewey FE, Gusarova V, Dunbar RL, O'Dushlaine C, Schurmann C, Gottesman O, McCarthy S, Van Hout CV, Bruse S, Dansky HM, Leader JB, Murray MF, Ritchie MD, Kirchner HL, Habegger L, Lopez A, Penn J, Zhao A, Shao W, Stahl N, Murphy AJ, Hamon S, Bouzelmat A, Zhang R, Shumel B, Pordy R, Gipe D, Herman GA, Sheu WHH, Lee IT, Liang KW, Guo X, Rotter JI, Chen YI, Kraus WE, Shah SH, Damrauer S, Small A, Rader DJ, Wulff AB, Nordestgaard BG, Tybjaerg-Hansen A, van den Hoek AM, Princen HMG, Ledbetter DH, Carey DJ, Overton JD, Reid JG, Sasiela WJ, Banerjee P, Shuldiner AR, Borecki IB, Teslovich TM, Yancopoulos GD, Mellis SJ, Gromada J and Baras A. Genetic and Pharmacologic Inactivation of ANGPTL3 and Cardiovascular Disease. N Engl J Med. 2017,377:211-221.
  4. Graham MJ, Lee RG, Brandt TA, Tai LJ, Fu W, Peralta R, Yu R, Hurh E, Paz E, McEvoy BW, Baker BF, Pham NC, Digenio A, Hughes SG, Geary RS, Witztum JL, Crooke RM and Tsimikas S. Cardiovascular and Metabolic Effects of ANGPTL3 Antisense Oligonucleotides. N Engl J Med. 2017,377:222-232.
  5. Gaudet D, Alexander VJ, Baker BF, Brisson D, Tremblay K, Singleton W, Geary RS, Hughes SG, Viney NJ, Graham MJ, Crooke RM, Witztum JL, Brunzell JD and Kastelein JJ. Antisense Inhibition of Apolipoprotein C-III in Patients with Hypertriglyceridemia. N Engl J Med. 2015,373:438-47.
  6. Lambert JE, Ramos-Roman MA, Browning JD and Parks EJ. Increased de novo lipogenesis is a distinct characteristic of individuals with nonalcoholic fatty liver disease. Gastroenterology. 2014,146:726-35.
  7. DeBose-Boyd RA and Ye J. SREBPs in Lipid Metabolism, Insulin Signaling, and Beyond. Trends Biochem Sci. 2018,43:358-368.
  8. Takeuchi Y, Yahagi N, Izumida Y, Nishi M, Kubota M, Teraoka Y, Yamamoto T, Matsuzaka T, Nakagawa Y, Sekiya M, Iizuka Y, Ohashi K, Osuga J, Gotoda T, Ishibashi S, Itaka K, Kataoka K, Nagai R, Yamada N, Kadowaki T and Shimano H. Polyunsaturated fatty acids selectively suppress sterol regulatory element-binding protein-1 through proteolytic processing and autoloop regulatory circuit. J Biol Chem. 2010,285:11681-91.
  9. Moon YA, Hammer RE and Horton JD. Deletion of ELOVL5 leads to fatty liver through activation of SREBP-1c in mice. J Lipid Res. 2009,50:412-23.
  10. Hannah VC, Ou J, Luong A, Goldstein JL and Brown MS. Unsaturated fatty acids down-regulate srebp isoforms 1a and 1c by two mechanisms in HEK-293 cells. J Biol Chem. 2001,276:4365-72.
  11. Kim H, Zhang H, Meng D, Russell G, Lee JN and Ye J. UAS domain of Ubxd8 and FAF1 polymerizes upon interaction with long-chain unsaturated fatty acids. J Lipid Res. 2013,54:2144-52.
  12. Botolin D, Wang Y, Christian B and Jump DB. Docosahexaenoic acid (22:6,n-3) regulates rat hepatocyte SREBP-1 nuclear abundance by Erk- and 26S proteasome-dependent pathways. J Lipid Res. 2006,47:181-92.
  13. Amemiya-Kudo M, Shimano H, Yoshikawa T, Yahagi N, Hasty AH, Okazaki H, Tamura Y, Shionoiri F, Iizuka Y, Ohashi K, Osuga J, Harada K, Gotoda T, Sato R, Kimura S, Ishibashi S and Yamada N. Promoter analysis of the mouse sterol regulatory element-binding protein-1c gene. J Biol Chem. 2000,275:31078-85.
  14. Hodson L, Bhatia L, Scorletti E, Smith DE, Jackson NC, Shojaee-Moradie F, Umpleby M, Calder PC and Byrne CD. Docosahexaenoic acid enrichment in NAFLD is associated with improvements in hepatic metabolism and hepatic insulin sensitivity: a pilot study. Eur J Clin Nutr. 2017,71:973-979.
  15. Donnelly KL, Smith CI, Schwarzenberg SJ, Jessurun J, Boldt MD and Parks EJ. Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. J Clin Invest. 2005,115:1343-51.
  16. Kim CW, Addy C, Kusunoki J, Anderson NN, Deja S, Fu X, Burgess SC, Li C, Ruddy M, Chakravarthy M, Previs S, Milstein S, Fitzgerald K, Kelley DE and Horton JD. Acetyl CoA Carboxylase Inhibition Reduces Hepatic Steatosis but Elevates Plasma Triglycerides in Mice and Humans: A Bedside to Bench Investigation. Cell Metab. 2017,26:394-406 e6.
  17. Yoshikawa T, Shimano H, Yahagi N, Ide T, Amemiya-Kudo M, Matsuzaka T, Nakakuki M, Tomita S, Okazaki H, Tamura Y, Iizuka Y, Ohashi K, Takahashi A, Sone H, Osuga Ji J, Gotoda T, Ishibashi S and Yamada N. Polyunsaturated fatty acids suppress sterol regulatory element-binding protein 1c promoter activity by inhibition of liver X receptor (LXR) binding to LXR response elements. J Biol Chem. 2002,277:1705-11.
  18. Pawar A, Botolin D, Mangelsdorf DJ and Jump DB. The role of liver X receptor-alpha in the fatty acid regulation of hepatic gene expression. J Biol Chem. 2003,278:40736-43.
  19. Zhao A, Yu J, Lew JL, Huang L, Wright SD and Cui J. Polyunsaturated fatty acids are FXR ligands and differentially regulate expression of FXR targets. DNA Cell Biol. 2004,23:519-26.
  20. Neuschwander-Tetri BA, Loomba R, Sanyal AJ, Lavine JE, Van Natta ML, Abdelmalek MF, Chalasani N, Dasarathy S, Diehl AM, Hameed B, Kowdley KV, McCullough A, Terrault N, Clark JM, Tonascia J, Brunt EM, Kleiner DE, Doo E and Network NCR. Farnesoid X nuclear receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic steatohepatitis (FLINT): a multicentre, randomised, placebo-controlled trial. Lancet. 2015,385:956-65.
  21. Bays HE, Braeckman RA, Ballantyne CM, Kastelein JJ, Otvos JD, Stirtan WG and Soni PN. Icosapent ethyl, a pure EPA omega-3 fatty acid: effects on lipoprotein particle concentration and size in patients with very high triglyceride levels (the MARINE study). J Clin Lipidol. 2012,6:565-72.
  22. Yokoyama M, Origasa H, Matsuzaki M, Matsuzawa Y, Saito Y, Ishikawa Y, Oikawa S, Sasaki J, Hishida H, Itakura H, Kita T, Kitabatake A, Nakaya N, Sakata T, Shimada K, Shirato K and Japan EPAlisI. Effects of eicosapentaenoic acid on major coronary events in hypercholesterolaemic patients (JELIS): a randomised open-label, blinded endpoint analysis. Lancet. 2007,369:1090-8.
  23. Wu S, Wu F, Ding Y, Hou J, Bi J and Zhang Z. Association of non-alcoholic fatty liver disease with major adverse cardiovascular events: A systematic review and meta-analysis. Sci Rep. 2016,6:33386.
  24. Scorletti E, Bhatia L, McCormick KG, Clough GF, Nash K, Hodson L, Moyses HE, Calder PC, Byrne CD and Study W. Effects of purified eicosapentaenoic and docosahexaenoic acids in nonalcoholic fatty liver disease: results from the Welcome* study. Hepatology. 2014,60:1211-21.
  25. Bhatia L, Scorletti E, Curzen N, Clough GF, Calder PC and Byrne CD. Improvement in non-alcoholic fatty liver disease severity is associated with a reduction in carotid intima-media thickness progression. Atherosclerosis. 2016,246:13-20.

Science News Commentaries

View All Science News Commentaries

-- The opinions expressed in this commentary are not necessarily those of the editors or of the American Heart Association --